Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where J. Shin is active.

Publication


Featured researches published by J. Shin.


European Heart Journal | 2015

Changes in ventricular remodelling and clinical status during the year following a single administration of stromal cell-derived factor-1 non-viral gene therapy in chronic ischaemic heart failure patients: the STOP-HF randomized Phase II trial

Eugene S. Chung; Leslie W. Miller; Amit N. Patel; Russell David Anderson; Farrell O. Mendelsohn; Jay H. Traverse; Kevin Silver; J. Shin; Gregory A. Ewald; Mary Jane Farr; Saif Anwaruddin; Francis Plat; Scott J. Fisher; Alexander T. AuWerter; Joseph M. Pastore; Rahul Aras; Marc S. Penn

Background Stromal cell-derived factor-1 (SDF-1) promotes tissue repair through mechanisms of cell survival, endogenous stem cell recruitment, and vasculogenesis. Stromal Cell-Derived Factor-1 Plasmid Treatment for Patients with Heart Failure (STOP-HF) is a Phase II, double-blind, randomized, placebo-controlled trial to evaluate safety and efficacy of a single treatment of plasmid stromal cell-derived factor-1 (pSDF-1) delivered via endomyocardial injection to patients with ischaemic heart failure (IHF). Methods Ninety-three subjects with IHF on stable guideline-based medical therapy and left ventricular ejection fraction (LVEF) ≤40%, completed Minnesota Living with Heart Failure Questionnaire (MLWHFQ) and 6-min walk distance (6 MWD), were randomized 1 : 1 : 1 to receive a single treatment of either a 15 or 30 mg dose of pSDF-1 or placebo via endomyocardial injections. Safety and efficacy parameters were assessed at 4 and 12 months after injection. Left ventricular functional and structural measures were assessed by contrast echocardiography and quantified by a blinded independent core laboratory. Stromal Cell-Derived Factor-1 Plasmid Treatment for Patients with Heart Failure was powered based on change in 6 MWD and MLWHFQ at 4 months. Results Subject profiles at baseline were (mean ± SD): age 65 ± 9 years, LVEF 28 ± 7%, left ventricular end-systolic volume (LVESV) 167 ± 66 mL, N-terminal pro brain natriuretic peptide (BNP) (NTproBNP) 1120 ± 1084 pg/mL, MLWHFQ 50 ± 20 points, and 6 MWD 289 ± 99 m. Patients were 11 ± 9 years post most recent myocardial infarction. Study injections were delivered without serious adverse events in all subjects. Sixty-two patients received drug with no unanticipated serious product-related adverse events. The primary endpoint was a composite of change in 6 MWD and MLWHFQ from baseline to 4 months follow-up. The primary endpoint was not met (P = 0.89). For the patients treated with pSDF-1, there was a trend toward an improvement in LVEF at 12 months (placebo vs. 15 mg vs. 30 mg ΔLVEF: −2 vs. −0.5 vs. 1.5%, P = 0.20). A pre-specified analysis of the effects of pSDF-1 based on tertiles of LVEF at entry revealed improvements in EF and LVESV from lowest-to-highest LVEF. Patients in the first tertile of EF (<26%) that received 30 mg of pSDF-1 demonstrated a 7% increase in EF compared with a 4% decrease in placebo (ΔLVEF = 11%, P = 0.01) at 12 months. There was also a trend towards improvement in LVESV, with treated patients demonstrating an 18.5 mL decrease compared with a 15 mL increase for placebo at 12 months (ΔLVESV = 33.5 mL, P = 0.12). The change in end-diastolic and end-systolic volume equated to a 14 mL increase in stroke volume in the patients treated with 30 mg of pSDF-1 compared with a decrease of −11 mL in the placebo group (ΔSV = 25 mL, P = 0.09). In addition, the 30 mg-treated cohort exhibited a trend towards improvement in NTproBNP compared with placebo at 12 months (−784 pg/mL, P = 0.23). Conclusions The blinded placebo-controlled STOP-HF trial demonstrated the safety of a single endocardial administration of pSDF-1 but failed to demonstrate its primary endpoint of improved composite score at 4 months after treatment. Through a pre-specified analysis the STOP-HF trial demonstrates the potential for attenuating LV remodelling and improving EF in high-risk ischaemic cardiomyopathy. The safety profile supports repeat dosing with pSDF-1 and the degree of left ventricular remodelling suggests the potential for improved outcomes in larger future trials.


Jacc-Heart Failure | 2015

The HVAD Left Ventricular Assist Device: Risk Factors for Neurological Events and Risk Mitigation Strategies.

Jeffrey J. Teuteberg; Mark S. Slaughter; Joseph G. Rogers; Edwin C. McGee; Francis D. Pagani; Robert J. Gordon; E. Rame; Michael A. Acker; Robert L. Kormos; Christopher T. Salerno; Thomas P. Schleeter; Daniel J. Goldstein; J. Shin; Randall C. Starling; Thomas C. Wozniak; Adnan S. Malik; Scott C. Silvestry; Gregory A. Ewald; Ulrich P. Jorde; Yoshifumi Naka; Emma J. Birks; Kevin B. Najarian; David R. Hathaway; Keith D. Aaronson; Advance Trial Investigators

OBJECTIVES The purpose of this study was to determine the risk factors for ischemic in hemorrhage cerebrovascular events in patients supported by the HeartWare ventricular assist device (HVAD). BACKGROUND Patients supported with left ventricular assist devices are at risk for both ischemic and hemorrhagic cerebrovascular events. METHODS Patients undergoing implantation with a HVAD as part of the bridge-to-transplant trial and subsequent continued access protocol were included. Neurological events (ischemic cerebrovascular accidents [ICVAs] and hemorrhagic cerebrovascular accidents [HCVAs]) were assessed, and the risk factors for these events were evaluated in a multivariable model. RESULTS A total of 382 patients were included: 140 bridge-to-transplant patients from the ADVANCE (Evaluation of the HeartWare Left Ventricular Assist Device for the Treatment of Advanced Heart Failure) clinical trial and 242 patients from the continued access protocol. Patients had a mean age of 53.2 years; 71.2% were male, and 68.1% were white. Thirty-eight percent had ischemic heart disease, and the mean duration of support was 422.7 days. The overall prevalence of ICVA was 6.8% (26 of 382); for HCVA, it was 8.4% (32 of 382). Pump design modifications and a protocol-driven change in the antiplatelet therapy reduced the prevalence of ICVA from 6.3% (17 of 272) to 2.7% (3 of 110; p = 0.21) but had a negligible effect on the prevalence of HVCA (8.8% [24 of 272] vs. 6.4% [7 of 110]; p = 0.69). Multivariable predictors of ICVA were aspirin ≤81 mg and atrial fibrillation; predictors of HCVA were mean arterial pressure >90 mm Hg, aspirin ≤81 mg, and an international normalized ratio >3.0. Eight of the 30 participating sites had established improved blood pressure management (IBPM) protocols. Although the prevalence of ICVA for those with and without IBPM protocols was similar (5.3% [6 of 114] vs. 5.2% [14 of 268]; p = 0.99), those with IBPM protocols had a significantly lower prevalence of HCVA (1.8% [2 of 114] vs. 10.8% [29 of 268]; p = 0.0078). CONCLUSIONS Anticoagulation, antiplatelet therapy, and blood pressure management affected the prevalence of cerebrovascular events after implantation of the HVAD. Attention to these clinical parameters can have a substantial impact on the occurrence of serious neurological events. (Evaluation of the HeartWare Left Ventricular Assist Device for the Treatment of Advanced Heart Failure [ADVANCE]; NCT00751972).


Circulation-heart Failure | 2015

Blood Pressure and Adverse Events During Continuous Flow Left Ventricular Assist Device Support

Omar Saeed; Rita Jermyn; Faraj Kargoli; Shivank Madan; Santhosh Mannem; Sampath Gunda; Cecilia Nucci; Sarah Farooqui; Syed Hassan; Allison J. McLarty; Michelle W. Bloom; Ronald Zolty; J. Shin; David A. D’Alessandro; D. Goldstein; Snehal R. Patel

Background—Adverse events (AEs), such as intracranial hemorrhage, thromboembolic event, and progressive aortic insufficiency, create substantial morbidity and mortality during continuous flow left ventricular assist device support yet their relation to blood pressure control is underexplored. Methods and Results—A multicenter retrospective review of patients supported for at least 30 days and ⩽18 months by a continuous flow left ventricular assist device from June 2006 to December 2013 was conducted. All outpatient Doppler blood pressure (DOPBP) recordings were averaged up to the time of intracranial hemorrhage, thromboembolic event, or progressive aortic insufficiency. DOPBP was analyzed as a categorical variable grouped as high (>90 mm Hg; n=40), intermediate (80–90 mm Hg; n=52), and controlled (<80 mm Hg; n=31). Cumulative survival free from an AE was calculated using Kaplan–Meier curves and Cox hazard ratios were derived. Patients in the high DOPBP group had worse baseline renal function, lower angiotensin-converting enzyme inhibitor or angiotensin receptor blocker usage during continuous flow left ventricular assist device support, and a more prevalent history of hypertension. Twelve (30%) patients in the high DOPBP group had an AE, in comparison with 7 (13%) patients in the intermediate DOPBP group and only 1 (3%) in the controlled DOPBP group. The likelihood of an AE increased in patients with a high DOPBP (adjusted hazard ratios [95% confidence interval], 16.4 [1.8–147.3]; P=0.012 versus controlled and 2.6 [0.93–7.4]; P=0.068 versus intermediate). Overall, a similar association was noted for the risk of intracranial hemorrhage (P=0.015) and progressive aortic insufficiency (P=0.078) but not for thromboembolic event (P=0.638). Patients with an AE had a higher DOPBP (90±10 mm Hg) in comparison with those without an AE (85±10 mm Hg; P=0.05). Conclusions—In a population at risk, higher DOPBP during continuous flow left ventricular assist device support was significantly associated with a composite of AEs.


Circulation-heart Failure | 2016

Antiplatelet Therapy and Adverse Hematologic Events During Heart Mate II Support

Omar Saeed; Aman M. Shah; Faraj Kargoli; Shivank Madan; Allison P. Levin; Snehal R. Patel; Rita Jermyn; Cesar Guerrero; J. Nguyen; Daniel B. Sims; J. Shin; David A. D’Alessandro; D. Goldstein; Ulrich P. Jorde

Background—Hematologic adverse events are common during continuous flow left ventricular assist device support; yet, their relation to antiplatelet therapy, including aspirin (ASA) dosing, is uncertain. Methods and Results—A single-center retrospective review of all patients supported by a continuous flow left ventricular assist device (Heart Mate II) from June 2006 to November 2014 was conducted. Patients were categorized into 3 groups: (1) ASA 81 mg+dipyridamole 75 mg daily (n=26) with a target international normalized ratio (INR) of 2 to 3 from June 2006 to August 2009; (2) ASA 81 mg daily (n=18) from September 2009 to August 2011 with a target INR of 1.5 to 2; and (3) ASA 325 mg daily from September 2011 to November 2014 with a target INR of 2 to 3 (n=70). Hemorrhagic and thrombotic outcomes were retrieved ⩽365 days after implantation. Cumulative survival free from adverse events was calculated using Kaplan–Meier curves and Cox proportional hazard ratios were generated. Hemorrhagic events occurred in 6 patients on ASA 81 mg+dipyridamole (26%; 0.42 events per patient year; mean INR at event, 2.2), 4 patients on ASA 81 mg (22%; 0.38 events per patient year; mean INR at event, 2.0), and in 38 patients on ASA 325 mg (54%; 1.4 events per patient year; mean INR at event, 2.2); P=0.004. Patients on ASA 325 mg had a higher adjusted hazard ratio of 2.9 (95% confidence interval, 1.2–7.0 versus ASA 81 mg+dipyridamole; P=0.02) and 3.4 (95% confidence interval, 1.2–9.5 versus ASA 81 mg; P=0.02) for hemorrhagic events. Thrombotic events rates were not different between groups. Conclusions—High-dose ASA in Heart Mate II patients treated concomitantly with warfarin is associated with an increased hazard of bleeding but does not reduce thrombotic events.


Circulation-heart Failure | 2017

Sildenafil Is Associated With Reduced Device Thrombosis and Ischemic Stroke Despite Low-Level Hemolysis on Heart Mate II Support

Omar Saeed; Sabarivinoth Rangasamy; Ibrahim Selevany; Shivank Madan; Jeremy Fertel; Ruth Eisenberg; Mohammad Aljoudi; Snehal R. Patel; J. Shin; Daniel B. Sims; Morayma Reyes Gil; Daniel J. Goldstein; Marvin J. Slepian; Henny H. Billett; Ulrich P. Jorde

Background Persistent low-level hemolysis (LLH) during continuous-flow mechanical circulatory support is associated with subsequent thrombosis. Free hemoglobin from ongoing hemolysis scavenges nitric oxide (NO) to create an NO deficiency which can augment platelet function leading to a prothrombotic state. The phosphodiesterase-5 inhibitor, sildenafil, potentiates NO signaling to inhibit platelet function. Accordingly, we investigated the association of sildenafil administration and thrombotic events in patients with LLH during Heart Mate II support. Methods and Results A single-center review of all patients implanted with a Heart Mate II who survived to discharge (n=144). LLH was defined by a discharge lactate dehydrogenase level of 400 to 700 U/L. Patients were categorized as (1) LLH not on sildenafil, (2) LLH on sildenafil, (3) no LLH not on sildenafil, and (4) no LLH on sildenafil. Age, sex, platelet count, and mean platelet volume were similar between groups. Seventeen patients had either device thrombosis or ischemic stroke. Presence of LLH was associated with a greater risk of thrombosis (adjusted hazard ratio, 15; 95% confidence interval, 4.5–50; P<0.001 versus no LLH, not on sildenafil). This risk was reduced in patients with LLH on sildenafil (adjusted hazard ratio, 1.7; 95% confidence interval, 0.2–16.1; P=0.61). Device thrombosis and ischemic stroke were associated with an increase in mean platelet volume (9.6±0.5 to 10.9±0.8 fL, P<0.001). Patients with LLH not on sildenafil had a greater increase in mean platelet volume in comparison to those with LLH on sildenafil (P<0.001). Conclusions Sildenafil is associated with reduced device thrombosis and ischemic stroke during ongoing LLH on Heart Mate II support.


Case reports in transplantation | 2014

Cardiac Failure after Liver Transplantation Requiring a Biventricular Assist Device

Rita Jermyn; Eiei Soe; David A. D'Alessandro; J. Shin; William Jakobleff; Daniel Schwartz; Milan Kinkhabwala; Paul J. Gaglio

Increased hepatic iron load in extrahepatic organs of cirrhotic patients with and without hereditary hemochromatosis portends a poorer long term prognosis after liver transplant. Hepatic as well as nonhepatic iron overload is associated with increased infectious and postoperative complications, including cardiac dysfunction. In this case report, we describe a cirrhotic patient with alpha 1 antitrypsin deficiency and nonhereditary hemochromatosis (non-HFE) that developed cardiogenic shock requiring mechanical circulatory support for twenty days after liver transplant. Upon further investigation, she was found to have significant iron deposition in both the liver and heart biopsies. Her heart regained complete and sustained recovery following ten days of mechanical biventricular support. This case highlights the importance of preoperatively recognizing extrahepatic iron deposition in patients referred for liver transplantation irrespective of etiology of liver disease as this may prevent postoperative complications.


Journal of Heart and Lung Transplantation | 2016

Severity of Hemolysis Is Associated with Death and Ischemic Stroke during Veno-Arterial Extracorporeal Membrane Support

Omar Saeed; W. Jakobleff; M. Chau; S. Rangasamy; M. Algodi; M. Makkiya; M. Cruz; Snehal R. Patel; Sandhya Murthy; Daniel B. Sims; J. Shin; D. Goldstein; Ulrich P. Jorde

Purpose: Acquired Von Willebrand Factor (vWF) deficiency due to the loss of high molecular weight multimers (HMWM) has been well documented during CF LVAD support. It has been proposed that lowering pump speed in response to clinical gastrointestinal bleeding (GIB) may decrease shear stress allowing for the return of HMWMs. In-vivo data supporting this practice is lacking. Methods: Subjects at least 30 days post implantation of a Heart Mate (HM) II were prospectively recruited from the LVAD clinic. After confirming INR was > 2.0, pump speed was decreased to 8000 rpm and maintained for 6 hours. Blood samples obtained at baseline and 6 hours were compared for 2 measures of acquired vWF deficiency: 1) the ristocetin cofactor activity to vWF antigen ratio (Rco:Ag) and 2) gel electrophoresis for vWF multimer distribution. Results: Four patients agreed to participation. They were 57±15 years old, all were male and had been on HM II support for 401±199 days. All patients tolerated speed reduction without any adverse events. At baseline speed, HMWMs were reduced in all 4 patients. After 6 hours at 8000 rpm, there was no change in the HMWM profile (Figure 1). Similarly, the Rco:Ag ratio was reduced (nl > 0.65) in 3 of 4 patients at baseline and did not significantly change after speed reduction (0.56 → 0.56, 0.74 → 0.67, 0.58 → 0.65, 0.47 → 0.45; p = 0.437). Conclusion: Decreasing pump speed during HM II support does not lead to restoration of HMW Von Willebrand multimers. These findings suggest there may be no benefit to speed reduction in response to GIB. ( 662)


Jacc-Heart Failure | 2017

Outcomes of Early Adolescent Donor Hearts in Adult Transplant Recipients

Shivank Madan; Snehal R. Patel; Peter Vlismas; Omar Saeed; Sandhya Murthy; S. Forest; W. Jakobleff; Daniel B. Sims; Jacqueline M. Lamour; Daphne T. Hsu; J. Shin; D. Goldstein; Ulrich P. Jorde


Journal of Heart and Lung Transplantation | 2014

Intracranial Hemorrhage Is Associated with a Higher Doppler Blood Pressure during Continuous Flow Left Ventricular Assist Device Support

Omar Saeed; Rita Jermyn; Santhosh Mannem; Cecilia Nucci; D. Casazza; S. Farooqui; Michelle W. Bloom; Allison J. McLarty; Ronald Zolty; J. Shin; D. D’Alessandro; D. Goldstein; Snehal R. Patel


Journal of Heart and Lung Transplantation | 2018

Neutrophil to Lymphocyte Ratio at the Time of LVAD Implant Predicts 30-day Readmission

N. Ahmed; H. Gandhi; Y. Kim; Omar Saeed; Snehal R. Patel; S. Murthy; J. Shin; S. Forrest; Daniel J. Goldstein; Ulrich P. Jorde; D.B. Sims

Collaboration


Dive into the J. Shin's collaboration.

Top Co-Authors

Avatar

Omar Saeed

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Snehal R. Patel

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ulrich P. Jorde

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

D. Goldstein

Montefiore Medical Center

View shared research outputs
Top Co-Authors

Avatar

Daniel B. Sims

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shivank Madan

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

D.B. Sims

Montefiore Medical Center

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Goldstein

Newark Beth Israel Medical Center

View shared research outputs
Top Co-Authors

Avatar

S. Forest

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

S. Murthy

Montefiore Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge